1
|
Qu P, Yuan J, Wu Y, Tian S, Wu Z, Chen P, Pan M, Weng H, Mai K, Zhang W. Yellow mealworm (Tenebrio molitor) meal replacing dietary fishmeal alters the intestinal microbiota, anti-oxidation and immunity of large yellow croaker (Larimichthyscrocea). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110272. [PMID: 40081435 DOI: 10.1016/j.fsi.2025.110272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The present study investigated the impact of dietary fishmeal replacement with yellow mealworm (Tenebrio molitor) meal (TM) on the anti-oxidation, immunity and intestinal microbiota of large yellow croaker (Larimichthys crocea), with an initial body weight of 189.18 ± 0.13 g. Seven isonitrogenous and isolipidic diets were made using TM replacing the fishmeal at 0 % (TM0), 15 % (TM15), 30 % (TM30), 45 % (TM45), 60 % (TM60), 75 % (TM75) and 100 % (TM100), respectively. Each experimental diet was randomly assigned to three replicate groups of large yellow croaker (100 fish per group). After an 80-day feeding trial, it was showed that the activities of maltase, lysozyme and acid phosphatase in intestine, as well as serum albumin concentration and total anti-oxidative capacity in serum of fish fed diets with TM replacing fishmeal more than 60 % (P < 0.05). The intestinal muscularis thickness and perimeter-to-diameter ratio decreased significantly in TM75 and TM100 groups compared to the control (P < 0.05). At the transcriptional level, the mRNA expression of occludin, zonula occludens-1 (zo-1), oligopeptide transporter 1 (pept1), and oligopeptide transporter 2 (pept2) in the intestine, along with nuclear factor erythroid 2-related factor 2 (nrf2) and interleukin-10 (il-10) in both the intestine and liver, were linearly downregulated as the fishmeal replacement level increased (P < 0.05). While the relative expression levels of kelch-like ECH-associated protein 1 (keap-1), nuclear factor-κB (nf-κb) and tumor necrosis factor-α (tnf-α) in the intestine and liver were linearly upregulated with increasing dietary fishmeal replacement levels (P < 0.05). Besides, increasing dietary fishmeal replacement levels maintained intestinal microbiota alpha diversity (P > 0.05), while altering the intestinal microbial composition (P < 0.05). In conclusion, replacing 45 % of fishmeal with TM (equivalent to 25.57 % TM in diet) had no significant negative effects on the intestinal microflora, anti-oxidation and immunity of large yellow croaker.
Collapse
Affiliation(s)
- Peng Qu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Jing Yuan
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Yang Wu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Shuangjie Tian
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Zhenhua Wu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Peng Chen
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Mingzhu Pan
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Huasong Weng
- Ningde Fufa Fisheries Co. Ltd, Ningde, 352100, China
| | - Kangsen Mai
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
2
|
Morgan MJ, Kim YS. RIPK3 in necroptosis and cancer. Mol Cells 2025; 48:100199. [PMID: 40010643 PMCID: PMC11938148 DOI: 10.1016/j.mocell.2025.100199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Receptor-interacting protein kinase-3 is essential for the cell death pathway called necroptosis. Necroptosis is activated by the death receptor ligands and pattern recognition receptors of the innate immune system, leading to significant consequences in inflammation and in diseases, particularly cancer. Necroptosis is highly proinflammatory compared with other modes of cell death because cell membrane integrity is lost, resulting in releases of cytokines and damage-associated molecular patterns that potentiate inflammation and activate the immune system. We discuss various ways that necroptosis is triggered along with its potential role in cancer and therapy.
Collapse
Affiliation(s)
- Michael J Morgan
- Department of Natural Sciences, Northeastern State University, Tahlequah, OK 74464, USA.
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Ajou University, Suwon 16499, Korea; Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
3
|
Bruni M, Lobefaro F, Pellegrini C, Mastrangelo M, Gualdi G, Esposito M, Antonetti P, De Sanctis P, Amerio P, Fargnoli MC. Psoriasis and cancer: the role of inflammation, immunosuppression, and cancer treatment. Expert Opin Biol Ther 2025; 25:395-411. [PMID: 40034077 DOI: 10.1080/14712598.2025.2471093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION The relationship between psoriasis, immunomodulatory therapies, and the risk of malignancies is complex and still debated. The scarcity of evidence in this field makes clinicians hesitate to prescribe biological therapies for 'difficult-to-treat' patients. AREAS COVERED Based on a comprehensive MEDLINE/PUBMED search of articles published up to November 2024, this review synthesizes the current evidence on the association between psoriasis and cancer. This review specifically addresses four key aspects: the overall cancer risk in psoriatic patients, the potential role of cytokines involved in psoriasis pathogenesis in tumor development, the association between biological therapies and the incidence of new malignancies in this population, and the risk of cancer recurrence or progression in patients with a history of malignancy who are treated with biologics. EXPERT OPINION Biological therapies do not significantly elevate malignancy risk compared to non-biological treatments or the general population. Evidence is also reassuring for patients with prior malignancy, showing no tumor progression or recurrence. These findings support the timely use of biological treatments in 'difficult-to-treat' patients. Regular cancer screenings and risk-factor minimization should always be recommended for psoriatic patients undergoing immunomodulatory therapies. Multidisciplinary management involving oncologists is suggested, particularly for patients with active and advanced oncological disease.
Collapse
Affiliation(s)
- Manfredo Bruni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Fabio Lobefaro
- Dermatology, Department of Medicine and Aging Science, University of Chieti-Pescara, Chieti, Italy
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Mirco Mastrangelo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giulio Gualdi
- Dermatology, Department of Medicine and Aging Science, University of Chieti-Pescara, Chieti, Italy
| | - Maria Esposito
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paolo Antonetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paolo De Sanctis
- Dermatology, Department of Medicine and Aging Science, University of Chieti-Pescara, Chieti, Italy
| | - Paolo Amerio
- Dermatology, Department of Medicine and Aging Science, University of Chieti-Pescara, Chieti, Italy
| | - Maria Concetta Fargnoli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
4
|
Xie XX, Sun JD, Zang MX, Zhang G, Li CX, Zhai XW, Shen W, Ge W, Cheng SF. LPA reduces the apoptosis of cryopreserved porcine skin-derived stem cells by inhibiting the regulatory factor TNF-α. Cryobiology 2025; 118:105189. [PMID: 39706283 DOI: 10.1016/j.cryobiol.2024.105189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Preserving the viability and functionality of stem cells during cryopreservation is crucial for their successful application in regenerative medicine. The aim of this study is to investigate the effect of lysophosphatidic acid (LPA) on reducing the apoptosis of cryopreserved porcine skin-derived stem cells (pSDSCs). Our findings revealed that LPA, at a concentration of 5 μM, significantly improved viability and reduced apoptosis in cryopreserved pSDSCs. Furthermore, our data indicated that LPA enters pSDSCs through receptor type 1 (LPAR1). In cryopreserved pSDSCs, after LPA treatment, the expression level of tumor necrosis factor alpha (TNF-α) protein decreased, suggesting TNF-α involvement in the regulation of the anti-apoptotic process. Additionally, we found that resiquimod (R848), a TNF-α activator, increased the level of apoptosis in cryopreserved pSDSCs. When cryopreserved pSDSCs were treated with both LPA and R848, the protective effect of LPA against apoptosis was decreased. In conclusion, our study demonstrates that LPA could effectively counteract the effect of TNF-α-induced apoptosis, thereby enhancing the survival rates of cryopreserved pSDSCs. Importantly, this study explored a novel mechanism of reducing apoptosis in cryopreserved stem cells.
Collapse
Affiliation(s)
- Xin-Xiang Xie
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jia-Dong Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ming-Xin Zang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Geng Zhang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chun-Xiao Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xiang-Wei Zhai
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Ge
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shun-Feng Cheng
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
5
|
Lilley T, Camera DM, Kwa FAA. Repairing muscle with broccoli-derived sulforaphane: A preclinical evaluation for the treatment of mitochondrial myopathies. Drug Discov Today 2025; 30:104283. [PMID: 39736463 DOI: 10.1016/j.drudis.2024.104283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Skeletal muscle health relies on the production of adenosine triphosphate (ATP) in the mitochondria. ATP production is accompanied by oxidative phosphorylation, which generates reactive oxygen species (ROS). When there is an imbalance in ROS levels, oxidative stress and subsequent mitochondrial dysfunction, mitochondrial myopathies including sarcopenia, chronic progressive external ophthalmoplegia, and proximal myopathy can result. Such incurable myopathies are characterised by aberrant metabolism, limited ATP production, and muscle atrophy. Broccoli-derived sulforaphane has emerged as a novel treatment for mitochondrial myopathies because of its antioxidant and anti-inflammatory properties. This review discusses preclinical studies that reveal sulforaphane's potential therapeutic benefits and limitations in treating mitochondrial myopathies.
Collapse
Affiliation(s)
- Thomas Lilley
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Donny M Camera
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Faith A A Kwa
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
6
|
Ni J, Huang S, Liang Z, Chen Z, Zhang S, Li G, An T. Concentration, pathogenic composition, and exposure risks of bioaerosol in large indoor public environments: A comparative study of urban and suburban areas. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177790. [PMID: 39615183 DOI: 10.1016/j.scitotenv.2024.177790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Biological contamination in larger indoor environments can lead to the outbreak of various infectious diseases. This study aimed to compare the pollution profiles and associated health risks of airborne microorganisms in different indoor settings between urban and suburban areas by culturing, sequencing, and toxicological evaluation. The results indicated that the average level of culturable bacteria was higher in urban areas (955 ± 259 CFU/m3) compared to suburban areas (850 ± 85 CFU/m3), with the highest concentrations found in the market (2170 ± 798 CFU/m3) and gymnasium (2010 ± 300 CFU/m3). Conversely, the total number of airborne bacteria was higher in classroom (2.09 × 105) and laboratory (1.95 × 105 copies/m3), likely due to the presence of viable but non-culturable cells. Additionally, the concentrations of 0.5-2.0 μm total particles were higher in the market and cafeteria. Dominant airborne genera included Acinetobacter and Pseudomonas for bacteria, Cladosporium and Aspergillus for fungi, as well as Geneviridae and Herpesviridae for viruses. Bacterial and viral diversity and richness were significantly higher in suburban areas compared to urban areas, with distinct viral communities observed in hospital. Cytotoxicity assays revealed lower viability of cells in response to bioaerosols from the library (52.3 %) and laboratory (54.5 %); while lower proliferation rates were found for the cells exposed to bioaerosol from gymnasium (5.4 %) and market (6.0 %), suggesting higher toxicity of these environments. Additionally, bioaerosol exposure may impair cellular innate immunity by increasing the expression of IL-6, IL-8, TNF-α, IFN-γ. Our findings provide valuable information for assessing and controlling bioaerosol-related health risks in indoor environments.
Collapse
Affiliation(s)
- Jiasheng Ni
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Simin Huang
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Zhishu Liang
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zhen Chen
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Simeng Zhang
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Guiying Li
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Taicheng An
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
7
|
Yahya MA, Alshammari GM, Osman MA, Al-Harbi LN, Alotaibi SN. Isoliquiritigenin Prevents the Development of Nephropathy by an HFD in Rats Through the Induction of Antioxidant Production and Inhibition of the MD-2/TLR4/NF-κB Pathway. BIOLOGY 2024; 13:984. [PMID: 39765652 PMCID: PMC11727570 DOI: 10.3390/biology13120984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/15/2025]
Abstract
This study tested the ISL against renal damage induced by a high-fat diet (HFD) and explored its underlying mechanisms. Adult male rats were assigned to four groups: (1) control on a standard diet (STD), (2) ISL on STD (30 mg/kg), (3) HFD, and (4) HFD + ISL (30 mg/kg). After 12 weeks of dietary intervention, ISL treatment led to significant reductions in body weight gain, visceral fat, and glucose and insulin levels in HFD-fed rats. Notably, ISL decreased serum urea and creatinine, increased serum albumin, and improved urinary profiles by lowering the urinary albumin and the albumin/creatinine ratio. Histological analyses revealed that ISL enhanced the glomerular structure and mitigated tubular damage, as evidenced by reduced urinary excretion of the kidney injury markers NGAL and KIM-1. Additionally, ISL significantly lowered cholesterol, triglycerides, and free fatty acids in both the control and HFD groups while also decreasing oxidized low-density lipoproteins (ox-LDLs) and malondialdehyde (MDA). Importantly, ISL enhanced renal antioxidant levels, increasing glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). Moreover, ISL downregulated mRNA levels of MD-2, Toll-like receptor-4 (TLR-4), and NF-κB, leading to reduced NF-κB p65 levels in renal tissues. In conclusion, ISL offers substantial protection against HFD-induced renal toxicity through mechanisms that attenuate metabolic stress, enhance antioxidant defenses, and inhibit the MD-2/TLR4/NF-κB inflammatory pathway.
Collapse
Affiliation(s)
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (M.A.Y.); (M.A.O.); (L.N.A.-H.); (S.N.A.)
| | | | | | | |
Collapse
|
8
|
Darand M, Golpour-Hamedani S, Karimi E, Hassanizadeh S, Mirzaei M, Arabi V, Nadjarzadeh A, Hosseinzadeh M. The association between adherence to unhealthy plant-based diet and risk of COVID-19: a cross-sectional study. BMC Infect Dis 2024; 24:1245. [PMID: 39501206 PMCID: PMC11536890 DOI: 10.1186/s12879-024-10115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The fast spread of the coronavirus disease 2019 (COVID-19) epidemic and its high mortality were quickly considered by the health community. Dietary patterns play an important role in strengthening or weakening the immune system and thus incidence of diseases. AIM The present study can provide a comprehensive picture of the association between adherence to unhealthy plant-based diet (uPDI) and COVID-19 incidence. METHODS This study was undertaken on 8157 adults' participants of the Yazd Health Study (YaHS) and Taghzieh Mardom-e-Yazd (TAMIZ) study aged 20 to 70 years. Data on dietary intakes were obtained using a validated food frequency questionnaire (FFQ). Multivariable logistic regression analysis was used to assess the association between uPDI and COVID-19. RESULTS We found a significant association between uPDI and the risk of COVID-19 (OR: 1.36; 95% CI: 1.05-1.75) in the crude model. After adjusting potential confounders, a significant increasing trend in the odds of COVID-19 across increasing quintiles of uPDI (OR: 1.58;95% CI: 1.05-2.37; P-value: 0.027) was observed. Stratified analysis based on sex indicated that uPDI significantly increased the risk of COVID-19 only in males (OR: 1.73;95% CI: 1.12-2.67; P-value: 0.012) and had no effect on females. CONCLUSIONS Participants in the highest quintiles of the uPDI had 58% higher odds of COVID-19 compared to subjects in the lowest quintile of uPDI. Although our study has promising results, stronger clinical studies are needed.
Collapse
Affiliation(s)
- Mina Darand
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Golpour-Hamedani
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Karimi
- Research Development Center, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Hassanizadeh
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Mirzaei
- Yazd Cardiovascular Research Centre, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vahid Arabi
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Nadjarzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Hosseinzadeh
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
9
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
10
|
Yang T, Xiang CG, Wang XH, Li QQ, Lei SY, Zhang KR, Ren J, Lu HM, Feng CL, Tang W. RIPK1 inhibitor ameliorates pulmonary injury by modulating the function of neutrophils and vascular endothelial cells. Cell Death Discov 2024; 10:152. [PMID: 38521771 PMCID: PMC10960796 DOI: 10.1038/s41420-024-01921-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
Acute lung injury (ALI) is an acute and progressive hypoxic respiratory failure that could progress to acute respiratory distress syndrome (ARDS) with a high mortality rate, thus immediate medical attention and supportive care are necessary. The pathophysiology of ALI is characterized by the disruption of the alveolar-capillary barrier and activation of neutrophils, leading to lung tissue damage. The receptor-interacting protein kinase 1 (RIPK1) has emerged as a promising target for the treatment of multiple inflammatory diseases, but the role of RIPK1 in the ALI remains poorly understood. In this study, we aimed to figure out the pathological role of RIPK1 in ALI, especially in the pulmonary immune microenvironment involving neutrophils and endothelial cells. In vivo experiments showed that RIPK1 inhibitor protected against lipopolysaccharide (LPS)-induced lung injury in mouse models, with reduced neutrophils and monocytes infiltration in the lungs. Further studies demonstrated that, besides the inhibitory action on necroptosis, RIPK1 inhibitor directly suppressed reactive oxygen species (ROS) generation and inflammatory cytokines secretion from neutrophils. Furthermore, RIPK1 inhibition maintains the barrier function in TNF-α-primed vascular endothelial cells and prevents their activation induced by the supernatant from LPS-stimulated neutrophils. Mechanistically, the aforementioned effects of RIPK1 inhibitor are associated with the NF-κB signaling pathway, which is partially independent of necroptosis inhibition. These results provide new evidence that RIPK1 inhibitor directly regulates the function of neutrophils and endothelial cells, as well as interferes with the interactions between these two cell types, therefore contributing to a better understanding of RIPK1 in ALI and providing a potential avenue for future therapeutic interventions.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cai-Gui Xiang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Han Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Qing Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shu-Yue Lei
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai-Rong Zhang
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Jing Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Hui-Min Lu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chun-Lan Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
11
|
Chi Z, Yang H, Liu J. Study on the combined toxicity of DEHP and lead on the blood system of rats. CHEMOSPHERE 2024; 349:140908. [PMID: 38072204 DOI: 10.1016/j.chemosphere.2023.140908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/01/2023] [Accepted: 12/03/2023] [Indexed: 01/10/2024]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a commonly used phthalate ester compound, while lead is a persistent and bioaccumulative heavy metal. Both can be exposed to the body through a variety of ways, which may have an impact on the blood system. In this study, we examined the impact of co-exposure to DEHP (0, 10, 100 mg/kg) and Pb (0, 5, 50 mg/kg) on the blood system of male SD rats. The study revealed that continuous exposure to DEHP and Pb for 20 days resulted in a decrease in leukocytes and lymphocytes, while an increase in neutrophils and monocytes. Co-exposure led to a significant decrease in the spleen coefficients. Furthermore, the combined exposure could increase the ratio of bone marrow cells in G1 phase, and decrease the ratio of cells in S phase and G2 phase. Cytokine testing showed that combined exposure affects the secretion of hematopoietic factors and may cause bone marrow cell apoptosis. Single or combined exposure to DEHP and Pb can cause oxidative stress in serum and bone marrow. Overall, these results indicate that the co-exposure of DEHP and Pb adversely affected the blood system of rats, mainly due to the induction of oxidative stress and ultimately affects the secretion of cytokines. The combined effect of the two substances is primarily antagonistic. These results have important implications for the risk assessment of combined pollution and provide valuable theoretical guidance.
Collapse
Affiliation(s)
- Zhenxing Chi
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Hangzhou, 310015, China.
| | - Hanfeng Yang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China
| | - Jia Liu
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, China
| |
Collapse
|
12
|
Sun X, Li F, Yuan L, Bing Z, Li X, Yang K. pH-responsive resveratrol-loaded ZIF-8 nanoparticles modified with tannic acid for promoting colon cancer cell apoptosis. J Biomed Mater Res B Appl Biomater 2024; 112:e35320. [PMID: 37702969 DOI: 10.1002/jbm.b.35320] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/30/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023]
Abstract
Resveratrol (Res) is known for its potential in treating various types of cancers, with a particular advantage of causing minimal toxic side effects. However, its clinical application is constrained by challenges such as poor bioavailability, low water solubility, and chemical instability in neutral and alkaline environments. In light of these limitations, we have developed a pH-responsive drug delivery nanoplatform, Res@ZIF-8/TA NPs, which exhibits good biocompatibility and shows promise for in vitro cancer therapy. Benefiting from the mild reaction conditions provided by zeolitic imidazolate frameworks (ZIFs), a "one-pot method" was used for drug synthesis and loading, resulting in a satisfactory loading capacity. Notably, Res@ZIF-8/TA NPs respond to acidic environments, leading to an improved drug release profile with a controlled release effect. Our cell-based experiments indicated that tannic acid (TA) modification enhances the biocompatibility of ZIFs. 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT assay), Hoechst 33342/PI staining, cell scratch assay, Transwell and Reverse Transcription quantitative PCR (RT-qPCR) assays further demonstrated that Res@ZIF-8/TA NPs inhibited colon cancer cell migration and invasion, and promoted apoptosis of colon cancer cells, suggesting a therapeutic potential and demonstrating anti-cancer properties. In conclusion, the Res@ZIF-8/TA NPs pH-responsive drug delivery systems we developed may offer a promising avenue for cancer therapy. By addressing some of the challenges associated with Res-based treatments, this system could contribute to advancements in cancer therapeutics.
Collapse
Affiliation(s)
- Xueqiang Sun
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Fuxin Li
- The People's Hospital of Hezhou Hepatobiliary, Pancreatic and Spleen Surgery, Hezhou, China
| | - Lingyan Yuan
- Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Zhitong Bing
- Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Xun Li
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Kehu Yang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
- Institution of Clinical Research and Evidence Based Medicine, Gansu Provincial Hospital, Lanzhou, China
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Geng S, Chen L, Lin W, Wan F, Le Z, Hu W, Chen H, Liu X, Huang Q, Zhang H, Lu JJ, Kong L. Exploring the Therapeutic Potential of Triptonide in Salivary Adenoid Cystic Carcinoma: A Comprehensive Approach Involving Network Pharmacology and Experimental Validation. Curr Pharm Des 2024; 30:2276-2289. [PMID: 38910414 DOI: 10.2174/0113816128315277240610052453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Salivary Adenoid Cystic Carcinoma (ACC) is characterized by a highly invasive and slow-growing pattern, and its etiology remains unidentified. Triptonide (TN) has demonstrated efficacy as a pharmacotherapeutic agent against ACC. Nonetheless, the specific targets and mechanism of molecular action underlying the effectiveness of TN in treating ACC have not been elucidated. OBJECTIVES By integrating network pharmacology within laboratory experiments, this research delves into the prospective targets and molecular mechanisms associated with the application of TN in treating ACC. METHODS Initially, pertinent targets associated with TN against ACC were acquired from public databases. Subsequently, a combination of network pharmacology and bioinformatics analysis was utilized to screen the top 10 hub targets and key signal pathways of TN-treating ACC. Finally, in vitro experiments involving various molecular assays were conducted to evaluate the biological phenotypes of cells following TN treatment, encompassing assessments of apoptosis levels, plate migration, and other parameters, thereby validating pivotal genes and pathways. RESULTS A total of 23 pertinent targets for TN in relation to ACC were identified, with the top 10 hub genes being MAPK8, PTGS2, RELA, MAPK14, NR3C1, HDAC1, PPARG, NFKBIA, AR, and PGR. There was a significant correlation between the TNF signaling pathway and the treatment of ACC with TN. In vitro experiments demonstrated that TN treatment elevated RELA phosphorylation while concurrently reducing MAPK14 phosphorylation and inducing G2/M arrest. TN exhibited the ability to enhance the apoptosis rate through increased caspase-3 activity, elevated levels of Reactive Oxygen Species (ROS), mitochondrial dysfunction, and inhibition of cell migration. CONCLUSION There is a potential therapeutic role for TN in the treatment of ACC through the activation of the TNF signaling pathway. Among the identified candidates, MAPK8, HDAC1, PTGS2, RELA, NR3C1, PPARG, NFKBIA, AR, and PGR emerge as the most pertinent therapeutic targets for TN in the context of ACC treatment.
Collapse
Affiliation(s)
- Shikai Geng
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Li Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Ziyu Le
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Huaiyuan Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Xingyu Liu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Qingting Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Haojiong Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| |
Collapse
|
14
|
Boran T, Zengin OS, Seker Z, Gunaydin Akyildiz A, Oztas E, Özhan G. The cyclin-dependent kinase inhibitor abemaciclib-induced hepatotoxicity: Insight on the molecular mechanisms in HepG2/THP-1 co-culture model. Toxicol Lett 2024; 391:1-12. [PMID: 37992977 DOI: 10.1016/j.toxlet.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Drug-induced liver injury (DILI) is one of the widespread causes of liver injury and immune system plays important role. Abemaciclib (ABE) is a cyclin-dependent kinase inhibitor used as monotherapy or combination therapy in the treatment of breast cancer. Like other kinase inhibitors, the underlying mechanisms of ABE-induced hepatotoxicity are not completely known yet. In the current study, hepatotoxicity of ABE was evaluated with HepG2/THP-1 co-culture model which has been developed in recent years for the evaluation of DILI potential. Following ABE treatment, oxidative stress, mitochondrial damage, cytokine secretion levels, apoptotic/necrotic cell death were determined. According to our results, ROS production along with GSH depletion was observed in HepG2 cells after ABE treatment. ABE promoted secretion of pro-inflammatory mediators (TNF-α and MCP-1) and declined anti-inflammatory cytokine IL-10 release. Besides, NFKβ and JNK1 protein expression levels increased following ABE treatment. ABE enhanced intracellular calcium levels, induced early apoptotic and necrotic cell deaths in HepG2 cells. Furthermore, the changes in some mitochondrial parameters including a reducement in intracellular ATP levels and complex V activity; hyperpolarized mitochondrial membrane potential and enhanced mitochondrial ROS levels were observed, whereas mitochondrial mass did not show any differences after ABE treatments. Therefore, ABE-induced hepatotoxic effects is probably via oxidative stress, inflammatory response and necrotic cell death rather than direct mitochondrial toxicity. In conclusion; the study makes a significant contribution to strengthening the infrastructure we have on in vitro toxicity mechanism evaluations, which are the basis of preclinical toxicity studies.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Istanbul University-Cerrahpasa, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34500 Istanbul, Turkey
| | - Ozge Sultan Zengin
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey
| | - Zehra Seker
- Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Aysenur Gunaydin Akyildiz
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Ezgi Oztas
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| | - Gül Özhan
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey.
| |
Collapse
|
15
|
Bagnol R, Siverino C, Barnier V, O'Mahony L, Grijpma DW, Eglin D, Moriarty TF. Physicochemical Characterization and Immunomodulatory Activity of Polyelectrolyte Multilayer Coatings Incorporating an Exopolysaccharide from Bifidobacterium longum. Biomacromolecules 2023; 24:5589-5604. [PMID: 37983925 DOI: 10.1021/acs.biomac.3c00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Immunoregulatory polysaccharides from probiotic bacteria have potential in biomedical engineering. Here, a negatively charged exopolysaccharide from Bifidobacterium longum with confirmed immunoregulatory activity (EPS624) was applied in multilayered polyelectrolyte coatings with positively charged chitosan. EPS624 and coatings (1, 5, and 10 layers and alginate-substituted) were characterized by the zeta potential, dynamic light scattering, size exclusion chromatography, scanning electron microscopy, and atomic force microscopy. Peripheral blood mononuclear cells (hPBMCs) and fibroblasts were exposed for 1, 3, 7, and 10 days with cytokine secretion, viability, and morphology as observations. The coatings showed an increased rugosity and exponential growth mode with an increasing number of layers. A dose/layer-dependent IL-10 response was observed in hPBMCs, which was greater than EPS624 in solution and was stable over 7 days. Fibroblast culture revealed no toxicity or metabolic change after exposure to EPS624. The EPS624 polyelectrolyte coatings are cytocompatible, have immunoregulatory properties, and may be suitable for applications in biomedical engineering.
Collapse
Affiliation(s)
- Romain Bagnol
- AO Research Institute Davos, Davos Platz 7270, Switzerland
- Technical Medical Centre, Department of Advanced Organ Engineering and Therapeutics, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, Enschede 7522 NB, The Netherlands
| | | | - Vincent Barnier
- UMR 5307 LGF, CNRS, Mines Saint-Etienne, Centre SMS, Saint-Etienne F-42023, France
| | - Liam O'Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland, University College Cork, Cork TH12 HW58, Ireland
| | - Dirk W Grijpma
- Technical Medical Centre, Department of Advanced Organ Engineering and Therapeutics, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, Enschede 7522 NB, The Netherlands
| | - David Eglin
- Technical Medical Centre, Department of Advanced Organ Engineering and Therapeutics, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, Enschede 7522 NB, The Netherlands
- Univ Jean Monnet, INSERM, Mines Saint-Étienne, U1059 Sainbiose, Saint-Étienne F-42023, France
| | | |
Collapse
|
16
|
Li H, Niu X, Zhang D, Qu MH, Yang K. The role of the canonical nf-κb signaling pathway in the development of acute liver failure. Biotechnol Genet Eng Rev 2023; 39:775-795. [PMID: 36578157 DOI: 10.1080/02648725.2022.2162999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
As a clinical emergency with a high mortality rate, the treatment of acute liver failure has been paid attention to by society. At present, liver transplantation is the most effective treatment for acute liver failure, but there is still an insufficient supply of liver sources and a poor prognosis. In view of the current therapeutic development of this disease, more researchers have turned their attention to the research of drugs related to the NF-κB pathway. The NF-κB canonical pathway has been proven to play a role in a variety of diseases, regulating inflammation, apoptosis, and other physiological processes. More and more evidence shows that the NF-κB canonical pathway regulates the pathogenesis of acute liver failure. In this review, we will summarize the regulation process of the NF-κB canonical pathway on acute liver failure, and develop a new way to treat acute liver failure by targeting the components of the pathway.
Collapse
Affiliation(s)
- Hanyue Li
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Xiao Niu
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Dajin Zhang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
| | - Mei-Hua Qu
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Kunning Yang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
| |
Collapse
|
17
|
Fan Z, Jia W. Lactobacillus casei-Derived Postbiotics Elevate the Bioaccessibility of Proteins via Allosteric Regulation of Pepsin and Trypsin and Introduction of Endopeptidases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37410960 DOI: 10.1021/acs.jafc.3c02125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
The potential of probiotics to benefit digestion has been widely reported, while its utilization in high-risk patients and potential adverse reactions have focused interest on postbiotics. A variable data-independent acquisition (vDIA)-based spatial-omics strategy integrated with unsupervised variational autoencoders was applied to profile the functional mechanism underlying the action of Lactobacillus casei-derived postbiotic supplementation in goat milk digestion in an infant digestive system, from a metabolomics-peptidomics-proteomics perspective. Amide and olefin derivatives were proved to elevate the activities of pepsin and trypsin through hydrogen bonding and hydrophobic forces based on allosteric effects, and recognition of nine endopeptidases and their cleavage to serine, proline, and aspartate were introduced by postbiotics, thereby promoting the generation of hydrophilic peptides and elevating the bioaccessibility of goat milk protein. The peptides originating from αs1-casein, β-casein, β-lactoglobulin, Ig-like domain-containing protein, κ-casein, and serum amyloid A protein, with multiple bioactivities including angiotensin I-converting enzyme (ACE)-inhibitory, osteoanabolic, dipeptidyl peptidase IV (DPP-IV) inhibitory, antimicrobial, bradykinin-potentiating, antioxidant, and anti-inflammatory activities, were significantly increased in the postbiotic supplementation group, which was also considered to potentially prevent necrotizing enterocolitis through inhibiting the multiplication of pathogenic bacteria and blocking signal transducer and activator of transcription 1 and nuclear factor kappa-light-chain-enhancer of activated B cells inflammatory pathways. This research deepened the understanding of the mechanism underlying the postbiotics affecting goat milk digestion, which established a critical groundwork for the clinical application of postbiotics in infant complementary foods.
Collapse
Affiliation(s)
- Zibian Fan
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, China
| |
Collapse
|
18
|
Gupta P, Singh A, Verma AK, Kant S, Pandey AK, Mishra A, Khare P, Prakash V. Nanoencapsulation of Docetaxel Induces Concurrent Apoptosis and Necroptosis in Human Oral Cancer Cells (SCC-9) via TNF-α/RIP1/RIP3 Pathway. Indian J Clin Biochem 2023; 38:351-360. [PMID: 37234186 PMCID: PMC10205939 DOI: 10.1007/s12291-022-01055-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/22/2022] [Indexed: 11/29/2022]
Abstract
Human oral squamous cell carcinoma is the sixth most frequent malignant cancer, with an unacceptably high death rate that affects people's health. Albeit, there are several clinical approaches for diagnosing and treating oral cancer they are still far from ideal. We previously synthesised and characterised the docetaxel nanoformulation (PLGA-Dtx) and discovered that docetaxel nanoencapsulation may suppress oral cancer cells. The goal of this study was to figure out the mechanism involved in the suppression of oral cancer cell proliferation. We discovered that PLGA-Dtx inhibited SCC-9 cell growth considerably as compared to free docetaxel (Dtx), and that the viability of SCC-9 cells treated with PLGA-Dtx was decreased dose-dependently. MTT assay showed that PLGA-Dtx selectively inhibited the growth of PBMCs from oral cancer patients while sparing PBMCs from normal healthy controls. Further, flow cytometry analysis showed that PLGA-Dtx induced apoptosis and necroptosis in SCC-9 cells. G2/M cell cycle arrest has been confirmed on exposure of PLGA-Dtx for 24 h in SCC-9 cells. Interestingly, western blot investigation found that PLGA-Dtx increased the amounts of necroptic proteins and apoptosis-related proteins more efficiently than Dtx. Furthermore, PLGA-Dtx was more effective in terms of ROS generation, and mitochondrial membrane potential depletion. Pretreatment with necroptosis inhibitor Nec-1 efficiently reversed the ROS production and further recover MMP caused by PLGA-Dtx. Overall, this study revealed a mechanistic model of therapeutic response for PLGA-Dtx in SCC-9 cells and proposed its potency by inducing cell death via activation of concurrent apoptosis and necroptosis in SCC-9 cells via TNF-α/RIP1/RIP3 and caspase-dependent pathway.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Arpita Singh
- Department of Pharmacology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh 226010 India
| | - Ajay Kumar Verma
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Anuj Kumar Pandey
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Anupam Mishra
- Department of Otorhinolaryngology & Head Neck Surgery, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Puneet Khare
- Flow Cytometry Facility, Central Instrumentation Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001 India
| | - Ved Prakash
- Department of Pulmonary & Critical Care Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| |
Collapse
|
19
|
Ozoani H, Ezejiofor AN, Okolo KO, Orish CN, Cirovic A, Cirovic A, Orisakwe OE. Zinc and selenium attenuate quaternary heavy metal mixture-induced testicular damage via amplification of the antioxidant system, reduction in metal accumulation, inflammatory and apoptotic biomarkers. Toxicol Res 2023; 39:497-515. [PMID: 37398573 PMCID: PMC10313602 DOI: 10.1007/s43188-023-00187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 07/04/2023] Open
Abstract
Heavy metals (HMs) such as cadmium (Cd), lead (Pb), arsenic (As), and mercury (Hg) are highly toxic elements. They are often found together in nature as a heavy metal mixture (HMM) and are known to contribute to subfertility/infertility as environmental pollutants. This study aims to evaluate the potential benefits of treating HMM-induced testicular pathophysiology with zinc (Zn) and/or selenium (Se). Six-week-old male Sprague Dawley rats were grouped into 5 (n = 7). The control group received deionized water, while the other groups were treated with PbCl2 (20 mg kg-1), CdCl2 (1.61 mg kg-1), HgCl2 (0.40 mg kg-1), and Na2AsO3 (10 mg kg-1) in deionized water for 60 days. Additionally, groups III to V received Zn, Se, and Zn/Se, respectively, for 60 days. The study evaluated testis weight, metal accumulation, sperm analysis, FSH, LH, testosterone, prolactin, oxidative stress, antioxidants, pro-inflammatory and apoptotic markers, and presented structural changes in the testis as micrographs. HMM caused a significant increase in testis weight, metal accumulation, prolactin, oxidative stress, and pro-inflammatory and apoptotic markers, while significantly decreasing semen analysis, FSH, LH, and testosterone. Histology showed decreased spermatogenesis and spermiogenesis, as evidenced by the structure of the germ cells and spermatids. However, Zn, Se, or both ameliorated and reversed some of the observed damages. This study provides further evidence for the mitigative potential of Zn, Se, or both in reversing the damage inflicted by HMM in the testis, and as a countermeasure towards improving HM-induced decrease in public health fecundity. Graphical abstract
Collapse
Affiliation(s)
- Harrison Ozoani
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Port Harcourt, Choba Nigeria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Science and Technology, Nsukka, Enugu State Nigeria
| | - Anthonet N. Ezejiofor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Port Harcourt, Choba Nigeria
| | - Kenneth O. Okolo
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Science and Technology, Nsukka, Enugu State Nigeria
| | - Chinna N. Orish
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, 5323, Port Harcourt, Choba Nigeria
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Orish E. Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323, Port Harcourt, Choba Nigeria
- Provictoire Research Institute, Port Harcourt, Rivers State Nigeria
| |
Collapse
|
20
|
Shu L, Yuan Z, Li F, Cai Z. Oxidative stress and valvular endothelial cells in aortic valve calcification. Biomed Pharmacother 2023; 163:114775. [PMID: 37116353 DOI: 10.1016/j.biopha.2023.114775] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/15/2023] [Accepted: 04/23/2023] [Indexed: 04/30/2023] Open
Abstract
Calcified aortic valve disease (CAVD) is a common cardiovascular disease in elderly individuals. Although it was previously considered a degenerative disease, it is, in fact, a progressive disease involving multiple mechanisms. Aortic valve endothelial cells, which cover the outermost layer of the aortic valve and are directly exposed to various pathogenic factors, play a significant role in the onset and progression of CAVD. Hemodynamic changes can directly damage the structure and function of valvular endothelial cells (VECs). This leads to inflammatory infiltration and oxidative stress, which promote the progression of CAVD. VECs can regulate the pathological differentiation of valvular interstitial cells (VICs) through NO and thus affect the process of CAVD. Under the influence of pathological factors, VECs can also be transformed into VICs through EndMT, and then the pathological differentiation of VICs eventually leads to the formation of calcification. This review discusses the role of VECs, especially the role of oxidative stress in VECs, in the process of aortic valve calcification.
Collapse
Affiliation(s)
- Li Shu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Zhen Yuan
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Zhejun Cai
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
21
|
Jensen M, Thorsen NW, Hallberg LAE, Hägglund P, Hawkins CL. New insight into the composition of extracellular traps released by macrophages exposed to different types of inducers. Free Radic Biol Med 2023; 202:97-109. [PMID: 36990299 DOI: 10.1016/j.freeradbiomed.2023.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Neutrophil extracellular trap (NET) release plays a key role in many chronic disease settings, including atherosclerosis. They are critical to innate immune defence, but also contribute to disease by promoting thrombosis and inflammation. Macrophages are known to release extracellular traps or "METs", but their composition and role in pathological processes are less well defined. In this study, we examined MET release from human THP-1 macrophages exposed to model inflammatory and pathogenic stimuli, including tumour necrosis factor α (TNFα), hypochlorous acid (HOCl) and nigericin. In each case, there was release of DNA from the macrophages, as visualized by fluorescence microscopy with the cell impermeable DNA binding dye SYTOX green, consistent with MET formation. Proteomic analysis on METs released from macrophages exposed to TNFα and nigericin reveals that they are composed of linker and core histones, together with a range of cytosolic and mitochondrial proteins. These include proteins involved in DNA binding, stress responses, cytoskeletal organisation, metabolism, inflammation, anti-microbial activity, and calcium binding. Quinone oxidoreductase in particular, was highly abundant in all METs but has not been reported previously in NETs. Moreover, there was an absence of proteases in METs in contrast to NETs. Some of the MET histones, contained post-translational modifications, including acetylation and methylation of Lys but not citrullination of Arg. These data provide new insight into the potential implications of MET formation in vivo and their contributions to immune defence and pathology.
Collapse
Affiliation(s)
- Mathias Jensen
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Nicoline W Thorsen
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Line A E Hallberg
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Per Hägglund
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
22
|
Bavachin and Corylifol A Improve Muscle Atrophy by Enhancing Mitochondria Quality Control in Type 2 Diabetic Mice. Antioxidants (Basel) 2023; 12:antiox12010137. [PMID: 36671000 PMCID: PMC9855061 DOI: 10.3390/antiox12010137] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes reduces muscle mass and function. Chronic inflammation and mitochondrial dysfunction play critical roles in muscle atrophy pathogenesis. Here, we investigated the effects of bavachin and corylifol A from Psoralea corylifolia L. seeds on muscle atrophy in dexamethasone-treated mice and in db/db mice. Bavachin and corylifol A enhanced muscle strength and muscle mass in dexamethasone-treated mice. In diabetic mice, they enhanced muscle strength and cross-sectional areas. Bavachin and corylifol A suppressed inflammatory cytokine (interleukin-6 and tumor necrosis factor-α) expression levels by downregulating nuclear factor-κB phosphorylation. They decreased the muscle atrophic factor (myostatin, atrogin-1, and muscle RING finger-1) expression levels. They activated the AKT synthetic signaling pathway and induced a switch from fast-type glycolytic fibers (type 2B) to slow-type oxidative fibers (types I and 2A). They increased mitochondrial biogenesis and dynamic factor (optic atrophy-1, mitofusin-1/2, fission, mitochondrial 1, and dynamin 1-like) expression levels via the AMP-activated protein kinase-peroxisome proliferator-activated receptor gamma coactivator 1-alpha signaling pathway. They also improved mitochondrial quality by upregulating the mitophagy factor (p62, parkin, PTEN-induced kinase-1, and BCL2-interacting protein-3) expression levels. Therefore, bavachin and corylifol A exert potential therapeutic effects on muscle atrophy by suppressing inflammation and improving mitochondrial function.
Collapse
|
23
|
Yang Zhou J. Innate immunity and early liver inflammation. Front Immunol 2023; 14:1175147. [PMID: 37205101 PMCID: PMC10187146 DOI: 10.3389/fimmu.2023.1175147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/30/2023] [Indexed: 05/21/2023] Open
Abstract
The innate system constitutes a first-line defence mechanism against pathogens. 80% of the blood supply entering the human liver arrives from the splanchnic circulation through the portal vein, so it is constantly exposed to immunologically active substances and pathogens from the gastrointestinal tract. Rapid neutralization of pathogens and toxins is an essential function of the liver, but so too is avoidance of harmful and unnecessary immune reactions. This delicate balance of reactivity and tolerance is orchestrated by a diverse repertoire of hepatic immune cells. In particular, the human liver is enriched in many innate immune cell subsets, including Kupffer cells (KCs), innate lymphoid cells (ILCs) like Natural Killer (NK) cells and ILC-like unconventional T cells - namely Natural Killer T cells (NKT), γδ T cells and Mucosal-associated Invariant T cells (MAIT). These cells reside in the liver in a memory-effector state, so they respond quickly to trigger appropriate responses. The contribution of aberrant innate immunity to inflammatory liver diseases is now being better understood. In particular, we are beginning to understand how specific innate immune subsets trigger chronic liver inflammation, which ultimately results in hepatic fibrosis. In this review, we consider the roles of specific innate immune cell subsets in early inflammation in human liver disease.
Collapse
Affiliation(s)
- Jordi Yang Zhou
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- *Correspondence: Jordi Yang Zhou,
| |
Collapse
|
24
|
Gene Expression Profiling Elucidates Cellular Responses to NCX4040 in Human Ovarian Tumor Cells: Implications in the Mechanisms of Action of NCX4040. Cancers (Basel) 2022; 15:cancers15010285. [PMID: 36612280 PMCID: PMC9818835 DOI: 10.3390/cancers15010285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
The nitric oxide donor, NCX4040 is a non-steroidal anti-inflammatory-NO donor and has been shown to be extremely cytotoxic to a number of human tumors, including ovarian tumors cells. We have found that NCX4040 is cytotoxic against both OVCAR-8 and its adriamycin-selected OVCAR-8 variant (NCI/ADR-RES) tumor cell lines. While the mechanism of action of NCX4040 is not entirely clear, we as well as others have shown that NCX4040 generates reactive oxygen species (ROS) and induces DNA damage in tumor cells. Recently, we have reported that NCX4040 treatment resulted in a significant depletion of cellular glutathione, and formation of both reactive oxygen and nitrogen species (ROS/RNS), resulting in oxidative stress in these tumor cells. Furthermore, our results indicated that more ROS/RNS were generated in OVCAR-8 cells than in NCI/ADR-RES cells due to increased activities of superoxide dismutase (SOD), glutathione peroxidase and transferases expressed in NCI/ADR-RES cells. Further studies suggested that NCX4040-induced cell death may be mediated by peroxynitrite formed from NCX4040 in cells. In this study we used microarray analysis following NCX4040 treatment of both OVCAR-8 and its ADR-resistant variant to identify various molecular pathways involved in NCX4040-induced cell death. Here, we report that NCX4040 treatment resulted in the differential induction of oxidative stress genes, inflammatory response genes (TNF, IL-1, IL-6 and COX2), DNA damage response and MAP kinase response genes. A mechanism of tumor cell death is proposed based on our findings where oxidative stress is induced by NCX4040 from simultaneous induction of NOX4, TNF-α and CHAC1 in tumor cell death.
Collapse
|
25
|
He W, Chen P, Chen Q, Cai Z, Zhang P. Cytokine storm: behind the scenes of the collateral circulation after acute myocardial infarction. Inflamm Res 2022; 71:1143-1158. [PMID: 35876879 PMCID: PMC9309601 DOI: 10.1007/s00011-022-01611-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022] Open
Abstract
At least 17 million people die from acute myocardial infarction (AMI) every year, ranking it first among causes of death of human beings, and its incidence is gradually increasing. Typical characteristics of AMI include acute onset and poor prognosis. At present, there is no satisfactory treatment, but development of coronary collateral circulation (CCC) can be key to improving prognosis. Recent research indicates that the levels of cytokines, including those related to promoting inflammatory responses and angiogenesis, increase after the onset of AMI. In the early phase of AMI, cytokines play a vital role in inducing development of collateral circulation. However, when myocardial infarction is decompensated, cytokine secretion increases greatly, which may induce a cytokine storm and worsen prognosis. Cytokines can regulate the activation of a variety of signal pathways and form a complex network, which may promote or inhibit the establishment of collateral circulation. We searched for published articles in PubMed and Google Scholar, employing the keyword "acute myocardial infarction", "coronary collateral circulation" and "cytokine storm", to clarify the relationship between AMI and a cytokine storm, and how a cytokine storm affects the growth of collateral circulation after AMI, so as to explore treatment methods based on cytokine agents or inhibitors used to improve prognosis of AMI.
Collapse
Affiliation(s)
- Weixin He
- Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Peixian Chen
- Zhujiang Hospital, Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Qingquan Chen
- Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Zongtong Cai
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China
| | - Peidong Zhang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, People's Republic of China.
| |
Collapse
|
26
|
Roles of RIPK3 in necroptosis, cell signaling, and disease. Exp Mol Med 2022; 54:1695-1704. [PMID: 36224345 PMCID: PMC9636380 DOI: 10.1038/s12276-022-00868-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 12/29/2022] Open
Abstract
Receptor-interacting protein kinase-3 (RIPK3, or RIP3) is an essential protein in the "programmed" and "regulated" cell death pathway called necroptosis. Necroptosis is activated by the death receptor ligands and pattern recognition receptors of the innate immune system, and the findings of many reports have suggested that necroptosis is highly significant in health and human disease. This significance is largely because necroptosis is distinguished from other modes of cell death, especially apoptosis, in that it is highly proinflammatory given that cell membrane integrity is lost, triggering the activation of the immune system and inflammation. Here, we discuss the roles of RIPK3 in cell signaling, along with its role in necroptosis and various pathways that trigger RIPK3 activation and cell death. Lastly, we consider pathological situations in which RIPK3/necroptosis may play a role.
Collapse
|
27
|
Chen Q, Yang C, Zhang Z, Wang Z, Chen Y, Rossi V, Chen W, Xin M, Su Z, Du J, Guo W, Hu Z, Liu J, Peng H, Ni Z, Sun Q, Yao Y. Unprocessed wheat γ-gliadin reduces gluten accumulation associated with the endoplasmic reticulum stress and elevated cell death. THE NEW PHYTOLOGIST 2022; 236:146-164. [PMID: 35714031 PMCID: PMC9544600 DOI: 10.1111/nph.18316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/07/2022] [Indexed: 06/02/2023]
Abstract
Along with increasing demands for high yield, elite processing quality and improved nutrient value in wheat, concerns have emerged around the effects of gluten in wheat-based foods on human health. However, knowledge of the mechanisms regulating gluten accumulation remains largely unexplored. Here we report the identification and characterization of a wheat low gluten protein 1 (lgp1) mutant that shows extremely low levels of gliadins and glutenins. The lgp1 mutation in a single γ-gliadin gene causes defective signal peptide cleavage, resulting in the accumulation of an excessive amount of unprocessed γ-gliadin and a reduced level of gluten, which alters the endoplasmic reticulum (ER) structure, forms the autophagosome-like structures, leads to the delivery of seed storage proteins to the extracellular space and causes a reduction in starch biosynthesis. Physiologically, these effects trigger ER stress and cell death. This study unravels a unique mechanism that unprocessed γ-gliadin reduces gluten accumulation associated with ER stress and elevated cell death in wheat. Moreover, the reduced gluten level in the lgp1 mutant makes it a good candidate for specific diets for patients with diabetes or kidney diease.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Changfeng Yang
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Zhaoheng Zhang
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Zihao Wang
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Yongming Chen
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Vincenzo Rossi
- Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial CropsI‐24126BergamoItaly
| | - Wei Chen
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan)Huazhong Agricultural UniversityWuhan430070China
| | - Mingming Xin
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Zhenqi Su
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Jinkun Du
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Weilong Guo
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Zhaorong Hu
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Jie Liu
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Huiru Peng
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Zhongfu Ni
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Qixin Sun
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| | - Yingyin Yao
- State Key Laboratory for Agrobiotechnology, Frontiers Science Center for Molecular Design Breeding, Key Laboratory of Crop Heterosis and Utilization (MOE), and Beijing Key Laboratory of Crop Genetic ImprovementChina Agricultural UniversityBeijing100193China
| |
Collapse
|
28
|
Deng Z, Wu N, Suo Q, Wang J, Yue Y, Geng L, Zhang Q. Fucoidan, as an immunostimulator promotes M1 macrophage differentiation and enhances the chemotherapeutic sensitivity of capecitabine in colon cancer. Int J Biol Macromol 2022; 222:562-572. [PMID: 36170928 DOI: 10.1016/j.ijbiomac.2022.09.201] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2023]
Abstract
Chemotherapy resistance is one of the most critical challenges in colorectal cancer (CRC) treatment. The occurrence and development of chemotherapy resistance closely related to the tumor immune microenvironment (TIME). As the most important immunosuppressive immune cells infiltrating into the TIME, macrophages are essential for chemotherapy resistance in CRC treatment. In this study, we found that a kind of fucoidan (FPS1M) induced macrophages differentiation to the M1 phenotype, and this transformation promoted cancer cells apoptosis both in vitro and in vivo. TNFα is a key mediator of FPS1M-induced tumorcidal activity of macrophages. Mechanistically, as a stimulator of TLR4, FPS1M enhanced macrophages glycolysis and regulated macrophages differentiation to the M1 phenotype by the activation of TLR4 mediated PI3K/AKT/mTOR signaling axis. In addition, FPS1M improved the immunosuppressed tumor microenvironment by increasing the infiltration of M1 macrophages in tumor tissue, which was conducive to improving the sensitivity of tumor to chemotherapy. Collectively, our findings demonstrated that FPS1M has the great potential to be used in tumor immunotherapy. The results also suggested that the combination of FPS1M with capecitabine is an alternative therapy method for colon cancer.
Collapse
Affiliation(s)
- Zhenzhen Deng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Wu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine drugs and biological products, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Qishan Suo
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Yang Yue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Lihua Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Quanbin Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China.
| |
Collapse
|
29
|
Babu S, Manoharan S, Ottappilakkil H, Perumal E. Role of oxidative stress-mediated cell death and signaling pathways in experimental fluorosis. Chem Biol Interact 2022; 365:110106. [PMID: 35985521 DOI: 10.1016/j.cbi.2022.110106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/03/2022]
Abstract
Free radicals and other oxidants have enticed the interest of researchers in the fields of biology and medicine, owing to their role in several pathophysiological conditions, including fluorosis (Fluoride toxicity). Radical species affect cellular biomolecules such as nucleic acids, proteins, and lipids, resulting in oxidative stress. Reactive oxygen species-mediated oxidative stress is a common denominator in fluoride toxicity. Fluorosis is a global health concern caused by excessive fluoride consumption over time. Fluoride alters the cellular redox homeostasis, and its toxicity leads to the activation of cell death mechanisms like apoptosis, autophagy, and necroptosis. Even though a surfeit of signaling pathways is involved in fluorosis, their toxicity mechanisms are not fully understood. Thus, this review aims to understand the role of reactive species in fluoride toxicity with an outlook on the effects of fluoride in vitro and in vivo models. Also, we emphasized the signal transduction pathways and the mechanism of cell death implicated in fluoride-induced oxidative stress.
Collapse
Affiliation(s)
- Srija Babu
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Harsheema Ottappilakkil
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
| |
Collapse
|
30
|
Devi K, Soni S, Tripathi V, Pandey R, Moharana B. Ethanolic Extract of Tridax procumbens Mitigates Pulmonary Inflammation via Inhibition of NF-κB/p65/ERK Mediated Signalling in an Allergic Asthma Model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154008. [PMID: 35263673 DOI: 10.1016/j.phymed.2022.154008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/22/2022] [Accepted: 02/16/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Tridax procumbens is a traditionally used medicinal plant with high content of active phytoconstituents having anti-inflammatory activity. Accumulating evidences have shown that Tridax procumbens efficaciously diminished oxidative stress and inflammation. However the anti-inflammatory role of Tridax procumbens is not obscured in allergic asthma. PURPOSE Aim of this study was to decipher the anti-inflammatory role of Tridax procumbens in allergic asthma and its underlying mechanism. METHODS Ethanolic extract of Tridax procumbens (TP) was prepared and major phytoconstituents (flavonoids) were characterized by biochemical and UPLC/MS analysis. Rats were sensitized and challenged with environmental allergen ovalbumin (OVA) and lipopolysaccharide (LPS) to establish an allergic asthma model. Persuasive anti-inflammatory role of TP was demonstrated in vivo (100, 200 and 400 mg/kg) and in vitro (250, 125, 75 and 25 µg/ml) experiments. RESULTS Characterization by UPLC/MS analysis showed the presence of various bioactive flavonoids. In in vitro study, significant reduction in ROS production, apoptosis and mitochondrial dysfunction were observed in alveolar type II cells upon pre-treatment with TP (250, 125, 75 and 25 µg/ml) in a concentration-dependant manner. In vivo, TP (200 mg/kg) oral administration showed robust anti-oxidative activity. TP treatment abrogated bronchial wall thickening, immune cell infiltration and bronchial wall fibre deposition. Immunohistochemical analysis showed the diminished expression of IL-1β, IL-6 in bronchial epithelium and vascular endothelium. TP abrogated inflammation by reducing the level of inflammatory cytokines including IL-2, IFN-γ, IL-6 and MCP-1, as well as inflammatory markers including TWEAK, TNF-α, TNF-R1 and its downstream transcription factor NF-ҡB/p65 activation and its nuclear translocation. Western blot analysis of TP treated lung tissue and alveolar type II cells showed reduced phosphorylation of ERK1/2 significantly. CONCLUSION TP exhibited anti-inflammatory activity by inhibition of ROS production and down-regulation of NF-ҡB/ERK signalling in vitro and in vivo asthma model. Thus, TP can be envisaged as an effective anti-inflammatory agent for OVA-induced allergic asthma.
Collapse
Affiliation(s)
- Kusum Devi
- Division of Pharmacology, CSIR-CDRI, Lucknow, India; Academy of Scientific & Innovative Research (AcSIR), Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - Sakshi Soni
- Division of Pharmacology, CSIR-CDRI, Lucknow, India
| | | | - Richa Pandey
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow, India
| | - Baisakhi Moharana
- Division of Pharmacology, CSIR-CDRI, Lucknow, India; Academy of Scientific & Innovative Research (AcSIR), Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
31
|
Lee J, Lee S, Min S, Kang SW. RIP3-Dependent Accumulation of Mitochondrial Superoxide Anions in TNF-α-Induced Necroptosis. Mol Cells 2022; 45:193-201. [PMID: 35289306 PMCID: PMC9001146 DOI: 10.14348/molcells.2021.0260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/29/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022] Open
Abstract
Excessive production of reactive oxygen species (ROS) is a key phenomenon in tumor necrosis factor (TNF)-α-induced cell death. However, the role of ROS in necroptosis remains mostly elusive. In this study, we show that TNF-α induces the mitochondrial accumulation of superoxide anions, not H2O2, in cancer cells undergoing necroptosis. TNF-α-induced mitochondrial superoxide anions production is strictly RIP3 expression-dependent. Unexpectedly, TNF-α stimulates NADPH oxidase (NOX), not mitochondrial energy metabolism, to activate superoxide production in the RIP3-positive cancer cells. In parallel, mitochondrial superoxide-metabolizing enzymes, such as manganese-superoxide dismutase (SOD2) and peroxiredoxin III, are not involved in the superoxide accumulation. Mitochondrial-targeted superoxide scavengers and a NOX inhibitor eliminate the accumulated superoxide without affecting TNF-α-induced necroptosis. Therefore, our study provides the first evidence that mitochondrial superoxide accumulation is a consequence of necroptosis.
Collapse
Affiliation(s)
- Jiyoung Lee
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul 03760, Korea
| | - Sunmi Lee
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul 03760, Korea
| | - Seongchun Min
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul 03760, Korea
| | - Sang Won Kang
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
32
|
Potential Pathophysiological Mechanisms Underlying Multiple Organ Dysfunction in Cytokine Release Syndrome. Mediators Inflamm 2022; 2022:7137900. [PMID: 35431655 PMCID: PMC9007670 DOI: 10.1155/2022/7137900] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022] Open
Abstract
In recent decades, many serious respiratory infections have broken out all over the world, including SARS-CoV, MERS, and COVID-19. They are characterized by strong infectivity, rapid disease progression, high mortality, and poor prognosis. Excessive immune system activation results in cytokine hypersecretion, which is an important reason for the aggravation of symptoms, and can spread throughout the body leading to systemic multiple organ dysfunction, namely, cytokine release syndrome (CRS). Although many diseases related to CRS have been identified, the mechanism of CRS is rarely mentioned clearly. This review is intended to clarify the pathogenetic mechanism of CRS in the deterioration of related diseases, describe the important signaling pathways and clinical pathophysiological characteristics of CRS, and provide ideas for further research and development of specific drugs for corresponding targets to treat CRS.
Collapse
|
33
|
Keller TCS, Lechauve C, Keller AS, Brooks S, Weiss MJ, Columbus L, Ackerman H, Cortese-Krott MM, Isakson BE. The role of globins in cardiovascular physiology. Physiol Rev 2022; 102:859-892. [PMID: 34486392 PMCID: PMC8799389 DOI: 10.1152/physrev.00037.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Globin proteins exist in every cell type of the vasculature, from erythrocytes to endothelial cells, vascular smooth muscle cells, and peripheral nerve cells. Many globin subtypes are also expressed in muscle tissues (including cardiac and skeletal muscle), in other organ-specific cell types, and in cells of the central nervous system (CNS). The ability of each of these globins to interact with molecular oxygen (O2) and nitric oxide (NO) is preserved across these contexts. Endothelial α-globin is an example of extraerythrocytic globin expression. Other globins, including myoglobin, cytoglobin, and neuroglobin, are observed in other vascular tissues. Myoglobin is observed primarily in skeletal muscle and smooth muscle cells surrounding the aorta or other large arteries. Cytoglobin is found in vascular smooth muscle but can also be expressed in nonvascular cell types, especially in oxidative stress conditions after ischemic insult. Neuroglobin was first observed in neuronal cells, and its expression appears to be restricted mainly to the CNS and the peripheral nervous system. Brain and CNS neurons expressing neuroglobin are positioned close to many arteries within the brain parenchyma and can control smooth muscle contraction and thus tissue perfusion and vascular reactivity. Overall, reactions between NO and globin heme iron contribute to vascular homeostasis by regulating vasodilatory NO signals and scavenging reactive species in cells of the mammalian vascular system. Here, we discuss how globin proteins affect vascular physiology, with a focus on NO biology, and offer perspectives for future study of these functions.
Collapse
Affiliation(s)
- T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Steven Brooks
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, Virginia
| | - Hans Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
34
|
Mongirdienė A, Skrodenis L, Varoneckaitė L, Mierkytė G, Gerulis J. Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies. Biomedicines 2022; 10:602. [PMID: 35327404 PMCID: PMC8945343 DOI: 10.3390/biomedicines10030602] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
With respect to structural and functional cardiac disorders, heart failure (HF) is divided into HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Oxidative stress contributes to the development of both HFrEF and HFpEF. Identification of a broad spectrum of reactive oxygen species (ROS)-induced pathways in preclinical models has provided new insights about the importance of ROS in HFrEF and HFpEF development. While current treatment strategies mostly concern neuroendocrine inhibition, recent data on ROS-induced metabolic pathways in cardiomyocytes may offer additional treatment strategies and targets for both of the HF forms. The purpose of this article is to summarize the results achieved in the fields of: (1) ROS importance in HFrEF and HFpEF pathophysiology, and (2) treatments for inhibiting ROS-induced pathways in HFrEF and HFpEF patients. ROS-producing pathways in cardiomyocytes, ROS-activated pathways in different HF forms, and treatment options to inhibit their action are also discussed.
Collapse
Affiliation(s)
- Aušra Mongirdienė
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, LT-50161 Kaunas, Lithuania
| | - Laurynas Skrodenis
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Leila Varoneckaitė
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Gerda Mierkytė
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| | - Justinas Gerulis
- Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
| |
Collapse
|
35
|
Redding MC, Pan JH, Kim YJ, Batish M, Trabulsi J, Lee JH, Kim JK. Apiaceous vegetables protect against acrolein-induced pulmonary injuries through modulating hepatic detoxification and inflammation in C57BL/6 male mice. J Nutr Biochem 2022; 101:108939. [PMID: 35016997 DOI: 10.1016/j.jnutbio.2022.108939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/12/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022]
Abstract
Acrolein (Acr) is a reactive aldehyde in the environment. Acr causes oxidative stress and a cascade of catalytic events and has, thereby, been associated with increased risk of pulmonary diseases. Whether apiaceous vegetables (API) consumption can prevent Acr-induced pulmonary toxicity has not yet been explored hence, we investigated the effects of API on Acr-induced pulmonary damages in C57BL/6J mice. The mice were assigned into either negative control [NEG group; American Institute of Nutrition (AIN)-93G diet only], positive control (POS group; AIN-93G+Acr) or API intervention group (API group; AIN-93G+21% API+Acr). After 1 week of dietary intervention, the POS and API mice were exposed to Acr (10 µmol/kg body weight/day) for 5 days. During the exposure period, assigned diets remained the same. Prominent indicators lung of toxicity of POS mice were found, including mucus accumulation, macrophage infiltration, and hemorrhage, all of which were ameliorated by the API. Serum and lung inflammation markers, such as a tumor necrosis factor alpha were also increased by Acr while reduced by API. In the liver, API upregulated expression of glutathione S-transferases, which enhanced the metabolism of Acr into water-soluble 3-hydroxypropyl mercapturic acid for excretion. This is consistent with observed reductions in serum Acr-protein adducts. Taken together, our results suggest that API may provide protection against Acr-induced pulmonary damages and inflammation via enhancement of the hepatic detoxification of Acr.
Collapse
Affiliation(s)
- Mersady C Redding
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA; School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Jeong Hoon Pan
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA; School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| | - Mona Batish
- Department of Medical and Molecular Sciences, University of Delaware, Newark, USA
| | - Jillian Trabulsi
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, South Korea.
| | - Jae Kyeom Kim
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA; School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
36
|
Maes M, Kubera M, Kotańska M. Aberrations in the Cross-Talks Among Redox, Nuclear Factor-κB, and Wnt/β-Catenin Pathway Signaling Underpin Myalgic Encephalomyelitis and Chronic Fatigue Syndrome. Front Psychiatry 2022; 13:822382. [PMID: 35599774 PMCID: PMC9120845 DOI: 10.3389/fpsyt.2022.822382] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/23/2022] [Indexed: 01/07/2023] Open
Abstract
There is evidence that chronic fatigue spectrum disorders (CFAS-Ds), including myalgic encephalomyelitis (ME), chronic fatigue syndrome (CFS), and chronic fatigue with physiosomatic symptoms including when due to comorbid medical disease, are characterized by neuroimmune and neuro-oxidative biomarkers. This study was performed to delineate the protein-protein interaction (PPI) network of CFAS-D and to discover the pathways, molecular patterns, and domains enriched in their PPI network. We performed network, enrichment, and annotation analyses using differentially expressed proteins and metabolics, which were established in patients with CFAS-D. The PPI network analysis revealed that the backbone of the highly connective CFAS-D network comprises NFKB1, CTNNB1, ALB, peroxides, NOS2, tumor necrosis factor (TNF), and interleukin-6 (IL-6) and that the network comprises interconnected immune-oxidative-nitrosative and Wnt/β-catenin subnetworks. Multiomics enrichment analysis shows that the CFAS-D network is highly significantly associated with cellular (antioxidant) detoxification, hydrogen peroxide metabolic process, peroxidase and oxidoreductase activity, interleukin-10 (IL-10) anti-inflammatory signaling and neurodegenerative canonical Wnt, the β-catenin complex, cadherin domains, cell-cell junctions and TLR2/4 pathways, and the transcription factors nuclear factor kappa B (NF-κB) and RELA. The top 10 DOID annotations of the CFAS-D network include four intestinal, three immune system disorders, cancer, and infectious disease. The custom Gene Ontology (GO) term annotation analysis revealed that the CFAS-D network is associated with a response to a toxic substance, lipopolysaccharides, bacterium, or virus. In conclusion, CFAS-D may be triggered by a variety of stimuli and their effects are mediated by aberrations in the cross-talks between redox, NF-κB, and Wnt/β-catenin signaling pathways leading to dysfunctions in multicellular organismal homeostatic processes.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.,IMPACT Strategic Research Center, Deakin University, Geelong, VIC, Australia
| | - Marta Kubera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Magdalena Kotańska
- Department of Pharmacological Screening, Medical College, Jagiellonian University, Kraków, Poland
| |
Collapse
|
37
|
A Emara A, H Mohamed M, S Nada E, A Hashem N, S Mahmoud E, M Abd-Elmonem A, Y Talab E, N Hameed A, M Dabbash O, Amir S, A Abd-Elgwad M, H Mohamed A, S Othman A, S Mansour M, A Ali A, A Hussein M. Astaxanthin Attenuates D-Galactosamine-Induced Pancreatic Injury by Activating Antioxidant Enzymes and Inhibiting VEGF-C Gene Expression. Pak J Biol Sci 2022; 25:191-200. [PMID: 35234009 DOI: 10.3923/pjbs.2022.191.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
<b>Background and Objective:</b> Astaxanthin (3,3'-dihydroxy-β-β-carotene-4,4'-dione) is a carotenoid, commonly found in marine environments has been reported to possess versatile biological properties including anti-inflammatory and antioxidant. In this study, the pancreatic protective effect of astaxanthin was investigated in D-Galactosamine-induced pancreas injury in rats. <b>Materials and Methods:</b> In this experimental study, MTT assay was used to determine cytotoxic effects of the Astaxanthin on pnc1 cells. A total of 30 adult albino rats divided into 5 groups, six rats in each. Group I was given an equal amount of distilled water, group II was received 400 mg kg<sup>1</sup> b.wt. D-galactosamine on 15th day, groups III-V were treated with astaxanthin (50 and 100 mg kg<sup>1</sup>) and/or silymarin (50 mg kg<sup>1</sup>) for 14 days + 400 mg kg<sup>1</sup> b.wt. D-galactosamine on the 15th day, respectively. <b>Results:</b> IC<sub>50 </sub>of Astaxanthin against the pnc1 cell line was 92.9 μg mL<sup>1</sup>. The daily oral administration of astaxanthin (50 and 100 mg kg<sup>1</sup>) as well as silymarin (50 mg kg<sup>1</sup>) for 14 days to rats treated with D-galactosamine resulted in a significant improvement in plasma AST, ALT, ALP as well as pancreatic TNF-α, IL-1β, IL-10, NO and VEGF-C gene expression. On the other hand, inducible oral administration of astaxanthin increased the activity of pancreatic GSH, SOD, GPx, GR, CAT and the level of TBARs in D-galactosamine-treated pancreatic of rats. Furthermore, Astaxanthin almost normalized these effects in pancreatic tissue histoarchitecture and MRI examination. <b>Conclusion:</b> The obtained results showed that Astaxanthin protected experimental animals against D-galactosamine-induced pancreatic injury through activation of antioxidant enzymes and IL-10 and inhibition of VEGF-C activation.
Collapse
|
38
|
Nelson VK, Pullaiah CP, Saleem Ts M, Roychoudhury S, Chinnappan S, Vishnusai B, Ram Mani R, Birudala G, Bottu KS. Natural Products as the Modulators of Oxidative Stress: An Herbal Approach in the Management of Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:161-179. [PMID: 36472822 DOI: 10.1007/978-3-031-12966-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most commonly diagnosed and frequently occurred cancer in the males globally. The current treatment strategies available to treat prostate cancer are not much effective and express various adverse effects. Hence, there is an urgent need to identify novel treatment that can improve patient outcome. From times immemorial, natural products are highly recognized for novel drug development for various diseases including cancer. Cancer cells generally maintain higher basal levels of reactive oxygen species (ROS) when compared to normal cells due to its high metabolic rate. However, initiation of excess intracellular ROS production can not be tolerated by the cancer cells and induce several cell death signals which are in contrast to normal cells. Therefore, small molecules of natural origin that induce ROS can potentially kill cancer cells in specific and provide a better opportunity to develop a novel drug therapy. In this review, we elaborated various classes of medicinal compounds and their mechanism of killing prostate cancer cells through direct or indirect ROS generation. This can generate a novel thought to develop promising drug candidate to treat prostate cancer patients.
Collapse
Affiliation(s)
- Vinod K Nelson
- Department of Pharmaceutical Chemistry, Raghavendra Institute of Pharmaceutical Education and Research (Autonomous), Anantapuramu, Andhra Pradesh, India.
| | - Chitikela P Pullaiah
- Department of Pharmacology, Siddha Central Research Institute, Central Council for Research in Siddha, Ministry of AYUSH, Chennai, Tamil Nadu, India
| | - Mohammed Saleem Ts
- College of Pharmacy, Riyadh ELM University, Riyadh, Kingdom of Saudi Arabia, Riyadh
| | | | - Sasikala Chinnappan
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia
| | - Beere Vishnusai
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Ravishankar Ram Mani
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia
| | - Geetha Birudala
- Faculty of Pharmacy, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Kavya Sree Bottu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| |
Collapse
|
39
|
Wang S, Sun X, Wang M, Deng Z, Niu B, Chen Q. Effect of roasted peanut allergen Ara h 3 protein on the sensitization of Caco-2 cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:5325-5336. [PMID: 33650104 DOI: 10.1002/jsfa.11180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Roasted peanut is widely loved as a kind of food with rich taste. However, peanut allergy is one of the major threats to human health, which affects about 5% of children and 1.4-2% of adults in the world. RESULTS To evaluate the sensitization mechanism of peanut allergen Ara h 3, Caco-2 cells as the model, which has the similar structure and function to differentiated small intestinal epithelial cells. Compared with Ara h 3-raw (purified from raw peanut) group, more significant results such as the inhibited Caco-2 cell viability and proliferation, the increased secretion of reactive oxygen species (ROS) and the decreased transepithelial electrical resistance were obtained in Ara h 3-roasted (purified from roasted peanut) group. Accordingly, oxidative stress and NF-κB signaling pathway were more imbalanced, which lead to the increased of thymic stromal lymphopoietin (TSLP), interleukin 6 (IL-6), IL-8 and monocyte chemotactic protein 1 (MCP-1). Then, the gene expression of tight junction proteins ZO-1, occludin and JAM-1 were reduced, which proved that the integrity of the Caco-2 monolayer barrier is severely damaged. CONCLUSION These finding identify the mechanisms of the allergenicity of roasted peanut allergy proteins are probably associated with intestinal uptake and cytokine dependent allergies. The aggravated allergic reaction might be caused by the increment of TSLP, IL-6, IL-8 and MCP-1 due to the activated NF-κB signaling pathway, and the enhanced transport of Ara h 3-roasted protein by Caco-2 monolayer. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shuo Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaodong Sun
- School of Medicine, Shanghai University, Shanghai, China
| | - Minjia Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhirui Deng
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
40
|
Upregulation of Bax, TNF-α and down-regulation of Bcl-2 in liver cancer cells treated with HL-7 and HL-10 peptides. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00800-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Redox Signaling and Sarcopenia: Searching for the Primary Suspect. Int J Mol Sci 2021; 22:ijms22169045. [PMID: 34445751 PMCID: PMC8396474 DOI: 10.3390/ijms22169045] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, the age-related decline in muscle mass and function, derives from multiple etiological mechanisms. Accumulative research suggests that reactive oxygen species (ROS) generation plays a critical role in the development of this pathophysiological disorder. In this communication, we review the various signaling pathways that control muscle metabolic and functional integrity such as protein turnover, cell death and regeneration, inflammation, organismic damage, and metabolic functions. Although no single pathway can be identified as the most crucial factor that causes sarcopenia, age-associated dysregulation of redox signaling appears to underlie many deteriorations at physiological, subcellular, and molecular levels. Furthermore, discord of mitochondrial homeostasis with aging affects most observed problems and requires our attention. The search for the primary suspect of the fundamental mechanism for sarcopenia will likely take more intense research for the secret of this health hazard to the elderly to be unlocked.
Collapse
|
42
|
Brandts I, Balasch JC, Gonçalves AP, Martins MA, Pereira ML, Tvarijonaviciute A, Teles M, Oliveira M. Immuno-modulatory effects of nanoplastics and humic acids in the European seabass (Dicentrarchus labrax). JOURNAL OF HAZARDOUS MATERIALS 2021; 414:125562. [PMID: 34030413 DOI: 10.1016/j.jhazmat.2021.125562] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/11/2021] [Accepted: 02/26/2021] [Indexed: 06/12/2023]
Abstract
Pernicious effects of plastic particles, emergent contaminants worldwide, have been described in different species. In teleost species, alterations of immune function after exposure to nanoplastics (NPs) have been reported, but the interaction with cortisol - hypothalamic-pituitary-adrenal (HPI) axis has not yet been explored. Furthermore, the role of dissolved organic matter on the effects of NPs is poorly known. Thus, the aims of this research were to assess if polystyrene NPs (PSNPs) acted as a stressor on juvenile European seabass (Dicentrarchus labrax), interfering with the immune response, as well as to elucidate if humic acids (HA) modulated the potential effects of PSNPs. A short-term exposure to PSNPs and HA elicited an immuno-modulatory response, with an activation of steroidogenic stress-related pathways. An upregulation of anti-inflammatory cytokine (il10, tgfb) and stress-related (mc2r, gr1) transcripts were observed after exposure to HA and PSNPs both individually and in co-exposure. No notable alteration of inflammatory markers was consistently found, which may reflect a protective anti-inflammatory effect of HA in the presence of PSNPs. Nevertheless, there seems to be a more complex interaction between both components. Overall, data show that understanding the interaction of NPs with dissolved organic substances is key to deciphering their environmental risks.
Collapse
Affiliation(s)
- I Brandts
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - J C Balasch
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - A P Gonçalves
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - M A Martins
- CICECO-Aveiro Institute of Materials, Department of Materials and Ceramic Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - M L Pereira
- CICECO-Aveiro Institute of Materials, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - A Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis INTERLAB-UMU, Regional Campus of International Excellence Mare Nostrum, University of Murcia, Espinardo, Murcia 30100, Spain
| | - M Teles
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - M Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
43
|
James PT, Ali Z, Armitage AE, Bonell A, Cerami C, Drakesmith H, Jobe M, Jones KS, Liew Z, Moore SE, Morales-Berstein F, Nabwera HM, Nadjm B, Pasricha SR, Scheelbeek P, Silver MJ, Teh MR, Prentice AM. The Role of Nutrition in COVID-19 Susceptibility and Severity of Disease: A Systematic Review. J Nutr 2021; 151:1854-1878. [PMID: 33982105 PMCID: PMC8194602 DOI: 10.1093/jn/nxab059] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/22/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Many nutrients have powerful immunomodulatory actions with the potential to alter susceptibility to coronavirus disease 2019 (COVID-19) infection, progression to symptoms, likelihood of severe disease, and survival. OBJECTIVE The aim was to review the latest evidence on how malnutrition across all its forms (under- and overnutrition and micronutrient status) may influence both susceptibility to, and progression of, COVID-19. METHODS We synthesized information on 13 nutrition-related components and their potential interactions with COVID-19: overweight, obesity, and diabetes; protein-energy malnutrition; anemia; vitamins A, C, D, and E; PUFAs; iron; selenium; zinc; antioxidants; and nutritional support. For each section we provide: 1) a landscape review of pertinent material; 2) a systematic search of the literature in PubMed and EMBASE databases, including a wide range of preprint servers; and 3) a screen of 6 clinical trial registries. All original research was considered, without restriction to study design, and included if it covered: 1) severe acute respiratory syndrome coronavirus (CoV) 2 (SARS-CoV-2), Middle East respiratory syndrome CoV (MERS-CoV), or SARS-CoV viruses and 2) disease susceptibility or 3) disease progression, and 4) the nutritional component of interest. Searches took place between 16 May and 11 August 2020. RESULTS Across the 13 searches, 2732 articles from PubMed and EMBASE, 4164 articles from the preprint servers, and 433 trials were returned. In the final narrative synthesis, we include 22 published articles, 38 preprint articles, and 79 trials. CONCLUSIONS Currently there is limited evidence that high-dose supplements of micronutrients will either prevent severe disease or speed up recovery. However, results of clinical trials are eagerly awaited. Given the known impacts of all forms of malnutrition on the immune system, public health strategies to reduce micronutrient deficiencies and undernutrition remain of critical importance. Furthermore, there is strong evidence that prevention of obesity and type 2 diabetes will reduce the risk of serious COVID-19 outcomes. This review is registered at PROSPERO as CRD42020186194.
Collapse
Affiliation(s)
- Philip T James
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Zakari Ali
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Bonell
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Carla Cerami
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Modou Jobe
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Kerry S Jones
- National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Zara Liew
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Sophie E Moore
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- Department of Women and Children's Health, King's College London, London, United Kingdom
| | - Fernanda Morales-Berstein
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen M Nabwera
- Department of International Public Health, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Behzad Nadjm
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Pauline Scheelbeek
- Department of Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Centre on Climate Change and Planetary Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Matt J Silver
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Megan R Teh
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew M Prentice
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
44
|
Moghadam ZM, Henneke P, Kolter J. From Flies to Men: ROS and the NADPH Oxidase in Phagocytes. Front Cell Dev Biol 2021; 9:628991. [PMID: 33842458 PMCID: PMC8033005 DOI: 10.3389/fcell.2021.628991] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
The cellular formation of reactive oxygen species (ROS) represents an evolutionary ancient antimicrobial defense system against microorganisms. The NADPH oxidases (NOX), which are predominantly localized to endosomes, and the electron transport chain in mitochondria are the major sources of ROS. Like any powerful immunological process, ROS formation has costs, in particular collateral tissue damage of the host. Moreover, microorganisms have developed defense mechanisms against ROS, an example for an arms race between species. Thus, although NOX orthologs have been identified in organisms as diverse as plants, fruit flies, rodents, and humans, ROS functions have developed and diversified to affect a multitude of cellular properties, i.e., far beyond direct antimicrobial activity. Here, we focus on the development of NOX in phagocytic cells, where the so-called respiratory burst in phagolysosomes contributes to the elimination of ingested microorganisms. Yet, NOX participates in cellular signaling in a cell-intrinsic and -extrinsic manner, e.g., via the release of ROS into the extracellular space. Accordingly, in humans, the inherited deficiency of NOX components is characterized by infections with bacteria and fungi and a seemingly independently dysregulated inflammatory response. Since ROS have both antimicrobial and immunomodulatory properties, their tight regulation in space and time is required for an efficient and well-balanced immune response, which allows for the reestablishment of tissue homeostasis. In addition, distinct NOX homologs expressed by non-phagocytic cells and mitochondrial ROS are interlinked with phagocytic NOX functions and thus affect the overall redox state of the tissue and the cellular activity in a complex fashion. Overall, the systematic and comparative analysis of cellular ROS functions in organisms of lower complexity provides clues for understanding the contribution of ROS and ROS deficiency to human health and disease.
Collapse
Affiliation(s)
- Zohreh Mansoori Moghadam
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
45
|
Morin decreases acrolein-induced cell injury in normal human hepatocyte cell line LO2. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
46
|
Akbari Z, Reisi P, Torkaman-Boutorabi A, Farahmandfar M. Effect of Pentoxifylline on Apoptotic-Related Gene Expression Profile, Learning and Memory Impairment Induced by Systemic Lipopolysaccharide Administration in the Rat Hippocampus. Int J Prev Med 2020; 11:151. [PMID: 33209221 PMCID: PMC7643573 DOI: 10.4103/ijpvm.ijpvm_170_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/29/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Inflammation is one of the effective factors, in the development of functional disorders of the nervous system. Pentoxifylline (PTX) has an inhibitory effect on inflammatory factors. Therefore the aim of this study was to evaluate the effect of PTX on learning, memory and expression of genes, involved in neuronal survival in the rat hippocampus, following systemic lipopolysaccharide (LPS) injection. Methods: Male rats were randomly divided into 5 groups of control, LPS and LPS + PTX, receiving doses of 10, 25 and 50 mg/kg of PTX, respectively. In LPS groups, LPS was injected (5 mg/kg; intraperitoneal), and after one week, rats received intraperitoneal PTX for 14 days, in the treatment groups. Learning and memory were evaluated by object location task (OLT) and novel object recognition (NOR). Then, the hippocampus was dissected in order to measure the expression of the associated genes. Results: The results showed that peripheral LPS injection caused significant damage (P < 0.01) to learning and memory with respect to controls, but PTX with doses of 10 and 50 mg/kg prevented these impairments. Results from reverse transcription polymerase chain reaction (RT-PCR) showed that LPS significantly increased the expression of Bax and TNF- α with respect to controls. PTX in the LPS + PTX group significantly increased the expression of Bcl-2, BAD and Caspase-3. Conclusions: Other than the increased Bcl-2 expression, PTX had no significant effect on the expression of other genes, therefore further studies are needed to find out how PTX improves the learning and memory impairments, following the peripheral inflammation.
Collapse
Affiliation(s)
- Zahra Akbari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,International Campuses, Tehran University of Medical Sciences, Tehran, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anahita Torkaman-Boutorabi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Farahmandfar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Wu L, Pan Y. Reactive oxygen species mediate TNF-α-induced inflammatory response in bone marrow mesenchymal cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 22:1296-1301. [PMID: 32128094 PMCID: PMC7038432 DOI: 10.22038/ijbms.2019.37893.9006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objectives It is generally believed that the inflammatory response in bone marrow mesenchymal stem cells (BMSCs) transplantation leads to poor survival and unsatisfactory effects, and is mainly mediated by cytokines, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α). In this study, we explored the mechanisms underlying the TNF-α-induced inflammatory response in BMSCs. Materials and Methods We treated BMSCs with TNF-α (1 and 10 ng/ml) for 5 days. The expression levels of key inflammatory mediators were evaluated by Real-time PCR. Intracellular ROS level was measured by using a 2, 7-dichlorofluorescein diacetate (DCF-DA). Results We found that TNF-α treatment dramatically increased the expression levels of some key inflammatory mediators, including IL-6, IL-1β, IFN-γ and transforming growth factor β (TGF-β). Moreover, TNF-α induced intracellular oxidative stress by elevating intracellular reactive oxygen species (ROS) level, which is due to the increase of lipid peroxidation, the reduction of antioxidant Glutathione (GSH) levels and the inhibition of many antioxidant enzyme activities in BMSCs. Interestingly, 5 µM curcumin, a ROS scavenger, dramatically lowered the TNF-α-induced inflammatory response in BMSCs. In addition, TNF-α induced the activation of extracellular-signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinases (JNK), p38 and their down-stream transcription factors nuclear factor kappa B (NF-κB) pathway. Conclusion ROS mediated the TNF-α-induced inflammatory response via MAPK and NF-κB pathway, and may provide a novel strategy to prevent the inflammatory-dependent impairments in BMSCs.
Collapse
Affiliation(s)
- Liuzhong Wu
- Department of Periodontics, School of Stomatology, China Medical University, Liaoning Province, China.,Department of Periodontics, Shenyang Stomatological Hospital, Shenyang, Liaoning Province, China
| | - Yaping Pan
- Department of Periodontics, School of Stomatology, China Medical University, Liaoning Province, China
| |
Collapse
|
48
|
He X, Song Y, Wang L, Xu J. Protective effect of pyrrolidine dithiocarbamate on isoniazid/rifampicin‑induced liver injury in rats. Mol Med Rep 2019; 21:463-469. [PMID: 31746430 DOI: 10.3892/mmr.2019.10817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/01/2019] [Indexed: 11/05/2022] Open
Abstract
Isoniazid (INH) and rifampicin (RIF) continue to be first line anti‑tuberculosis (TB) drugs. However, the use of these drugs is associated with hepatotoxicity. Nuclear factor‑κB (NF‑κB) plays a crucial role in regulating immunity and inflammation. It has been reported that pyrrolidine dithiocarbamate (PDTC), an inhibitor of NF‑κB, exerts a hepatoprotective effect on acute and chronic liver damage. The aim of the present study was to explore the INH/RIF‑induced protective effects and mechanisms of PDTC on liver injury. Rats were intragastrically administered INH (50 mg/kg/day) and RIF (50 mg/kg/day) daily for 28 days. PDTC (50 mg/kg/day) was intraperitoneally injected 2 h after the co‑administration of INH and RIF to compare liver biochemical indicators in the serum, histopathological damage, NF‑κB activity, oxidative stress, hepatic mRNA expression of tumor necrosis factor (TNF)‑α, bile salt export pump (BSEP), and protein expression of BSEP. It was found that the inhibition of NF‑κB activation by PDTC treatment markedly alleviated liver biochemical and histological injury, decreased oxidative stress and mRNA levels of TNF‑α, and prevented decreases in BSEP mRNA and protein expression induced by the co‑administration of INH and RIF. Collectively, the present data suggested that INH/RIF‑induced liver injury is dependent on the activation of NF‑κB. PDTC exerted a therapeutic effect on INH/RIF‑induced liver injury by increasing BSEP expression, and exhibiting antioxidant and anti‑inflammatory activities.
Collapse
Affiliation(s)
- Xue He
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yulin Song
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Li Wang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Jianming Xu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
49
|
Abstract
Significance: Redox homeostasis is finely tuned and governed by distinct intracellular mechanisms. The dysregulation of this either by external or internal events is a fundamental pathophysiologic base for many pulmonary diseases. Recent Advances: Based on recent discoveries, it is increasingly clear that cellular redox state and oxidation of signaling molecules are critical modulators of lung disease and represent a final common pathway that leads to poor respiratory outcomes. Critical Issues: Based on the wide variety of stimuli that alter specific redox signaling pathways, improved understanding of the disease and patient-specific alterations are needed for the development of therapeutic targets. Further Directions: For the full comprehension of redox signaling in pulmonary disease, it is essential to recognize the role of reactive oxygen intermediates in modulating biological responses. This review summarizes current knowledge of redox signaling in pulmonary development and pulmonary vascular disease.
Collapse
Affiliation(s)
- Gaston Ofman
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Trent E Tipple
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
50
|
Martínez MA, Rodríguez JL, Lopez-Torres B, Martínez M, Martínez-Larrañaga MR, Anadón A, Ares I. Oxidative stress and related gene expression effects of cyfluthrin in human neuroblastoma SH-SY5Y cells: Protective effect of melatonin. ENVIRONMENTAL RESEARCH 2019; 177:108579. [PMID: 31330490 DOI: 10.1016/j.envres.2019.108579] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 06/10/2023]
Abstract
This study was designed to assess oxidative stress induction in human neuroblastoma SH-SY5Y cells in response to cyfluthrin exposure. Cell viability MTT assay was carried out to assess cyfluthrin cytotoxicity; IC30 and IC50 values for cyfluthrin were calculated to be 4.81 ± 0.92 μM and 19.39 ± 3.44 μM, respectively. Cyfluthrin induced a significant increase in ROS generation, lipid peroxides measured as malondialdehyde (MDA) and nitric oxide (NO) production and a significant decrease in NQO1 activity. The antioxidant activity of melatonin (MEL), Trolox, N-acetylcysteine (NAC) and Sylibin against cyfluthrin-induced oxidative stress was examined. Cyfluthrin increased significantly gene expressions of apoptosis, proinflammation and oxidative stress (Bax, Bcl-2, Casp-3, BNIP3, AKT1, p53, APAF1, NFκB1, TNFα and Nrf2) mediators. In the most genes, the mRNA levels induced by cyfluthrin were partially reduced by MEL (1 μM). Cyfluthrin effects on gene expression profiling of oxidative stress pathway by Real-Time PCR array analysis showed that of the 84 genes examined, (fold change > 1.5) changes in mRNA levels were detected in 31 genes: 13 upregulated and 18 down-regulated. A fold change >3.0 fold was observed on upregulated CYBB, DUOX1, DUOX2, AOX1, BNIP3, HSPA1A, NOS2, and NQO1 genes. The greater fold change reversion (2.5 fold) by MEL (1 μM) was observed on cyfluthrin-upregulated CYBB, AOX1, BNIP3 and NOS2 genes. These results demonstrated that oxidative stress is a key element in cyfluthrin induced neurotoxicity as well as MEL may play a role in reducing cyfluthrin-induced oxidative stress.
Collapse
Affiliation(s)
- María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - José-Luis Rodríguez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|